04.07.2014 Views

Innate immune response to viral infection Cytokine - sepeap

Innate immune response to viral infection Cytokine - sepeap

Innate immune response to viral infection Cytokine - sepeap

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

ARTICLE IN PRESS<br />

Cy<strong>to</strong>kine xxx (2008) xxx–xxx<br />

Contents lists available at ScienceDirect<br />

Cy<strong>to</strong>kine<br />

journal homepage: www.elsevier.com/locate/issn/10434666<br />

Review Article<br />

<strong>Innate</strong> <strong>immune</strong> <strong>response</strong> <strong>to</strong> <strong>viral</strong> <strong>infection</strong><br />

Shohei Koyama a,b , Ken J. Ishii a,c , Cevayir Coban a,b , Shizuo Akira a,b, *<br />

a Labora<strong>to</strong>ry of Host Defense, WPI Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan<br />

b Department of Host Defense, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan<br />

c Department of Molecular Pro<strong>to</strong>zoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan<br />

article<br />

info<br />

abstract<br />

Article his<strong>to</strong>ry:<br />

Received 2 June 2008<br />

Accepted 9 June 2008<br />

Available online xxxx<br />

Keywords:<br />

Pattern recognition recep<strong>to</strong>rs<br />

Type I interferons<br />

Nucleic acids<br />

In <strong>viral</strong> <strong>infection</strong>s the host innate <strong>immune</strong> system is meant <strong>to</strong> act as a first line defense <strong>to</strong> prevent <strong>viral</strong><br />

invasion or replication before more specific protection by the adaptive <strong>immune</strong> system is generated. In<br />

the innate <strong>immune</strong> <strong>response</strong>, pattern recognition recep<strong>to</strong>rs (PRRs) are engaged <strong>to</strong> detect specific <strong>viral</strong><br />

components such as <strong>viral</strong> RNA or DNA or <strong>viral</strong> intermediate products and <strong>to</strong> induce type I interferons<br />

(IFNs) and other pro-inflamma<strong>to</strong>ry cy<strong>to</strong>kines in the infected cells and other <strong>immune</strong> cells. Recently these<br />

innate <strong>immune</strong> recep<strong>to</strong>rs and their unique downstream pathways have been identified. Here, we summarize<br />

their roles in the innate <strong>immune</strong> <strong>response</strong> <strong>to</strong> virus <strong>infection</strong>, discrimination between self and <strong>viral</strong><br />

nucleic acids and inhibition by virulent fac<strong>to</strong>rs and provide some recent advances in the coordination<br />

between innate and adaptive <strong>immune</strong> activation.<br />

Ó 2008 Published by Elsevier Ltd.<br />

1. Introduction<br />

* Corresponding author. Address: Labora<strong>to</strong>ry of Host Defense, WPI Immunology<br />

Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-<br />

0871, Japan. Fax: +81 6 6879 8305.<br />

E-mail address: sakira@biken.osaka-u.ac.jp (S. Akira).<br />

All living organisms have developed several kinds of mechanisms<br />

<strong>to</strong> protect themselves from invasion by exogenous microorganisms,<br />

including viruses. Although the production of neutralizing antibodies<br />

and activation of cy<strong>to</strong><strong>to</strong>xic T lymphocytes (CTL) or natural killer<br />

(NK) cells are essential for a specific and effective anti<strong>viral</strong> <strong>immune</strong><br />

<strong>response</strong>, other host cells also possess some <strong>immune</strong> mechanism <strong>to</strong><br />

prevent <strong>viral</strong> <strong>infection</strong>.<br />

Although multiple cy<strong>to</strong>kines and chemokines are produced by<br />

several kinds of host cells in <strong>viral</strong> <strong>infection</strong>, type I IFNs are the principal<br />

cy<strong>to</strong>kines involved in the anti<strong>viral</strong> <strong>response</strong>. Type I IFNs<br />

include multiple IFN-a isoforms, a single IFN-b, and other members,<br />

such as IFN-e, -j, -x and so on [1]. In contrast <strong>to</strong> type II IFN<br />

(IFN-c), which is exclusively produced by T cells and NK cells, type<br />

I IFNs can be produced by all nucleated cells in <strong>response</strong> <strong>to</strong> virus<br />

<strong>infection</strong>. Type III IFNs, comprised of IFN-k1, k2 and k3, have also<br />

recently been identified [2]. These IFNs each have different recep<strong>to</strong>rs<br />

but share downstream signaling molecules and regulate the<br />

same genes. IFNs have pleiotropic functions. They increase the<br />

expression of intrinsic proteins including TRIM5a, Fv, Mx, eIF2a<br />

and 2 0 –5 0 OAS, and induce apop<strong>to</strong>sis of virus-infected cells and cellular<br />

resistance <strong>to</strong> <strong>viral</strong> <strong>infection</strong> [3]. In addition they activate NK<br />

cells and dendritic cells (DC) and induce the activation of the adaptive<br />

<strong>immune</strong> system [4]. The expression of type I IFN and cy<strong>to</strong>kine<br />

genes is regulated by an intracellular signaling pathway that is<br />

activated by germline-encoded PRRs. These recep<strong>to</strong>rs recognize<br />

molecular patterns specific <strong>to</strong> microorganisms, such as <strong>viral</strong> genome<br />

nucleic acids. Nucleic acids such as DNA and RNA are essential<br />

components of all living organisms, so discrimination between self<br />

and non-self nucleic acids is essential especially in virus <strong>infection</strong>.<br />

Recent advances in research in<strong>to</strong> innate immunity have revealed<br />

that this discrimination relies, <strong>to</strong> a great extent, on PRRs including<br />

Toll-like recep<strong>to</strong>rs (TLRs), retinoic acid-inducible gene I (RIG-I)-like<br />

recep<strong>to</strong>rs (RLRs), and nucleotide-binding oligomerization domain<br />

(NOD)-like recep<strong>to</strong>rs (NLRs). Here, we review the current understanding<br />

of innate <strong>immune</strong> recognition of viruses and discrimination<br />

between self and <strong>viral</strong> nucleic acids, and provide some<br />

recent advances in coordination between innate <strong>immune</strong> signaling<br />

and adaptive <strong>immune</strong> activation.<br />

2. <strong>Innate</strong> <strong>immune</strong> recep<strong>to</strong>rs for virus sensing<br />

2.1. Endosomal TLRs in DCs<br />

Several kinds of viruses utilize host endocytic pathways at the<br />

cell entry phase or budding, so they are inevitably surveyed by<br />

endosomal innate <strong>immune</strong> sensors. Endosomal TLRs, including<br />

TLR3, TLR7, TLR8 and TLR9, share the property of being activated<br />

by nucleic acids. Their expression can be increased by type I IFNs<br />

but their distribution is restricted. TLR7 and TLR9 are highly<br />

expressed in plasmacy<strong>to</strong>id DCs (pDCs) which are expert cells<br />

known <strong>to</strong> produce a large amount of type I IFNs in <strong>response</strong> <strong>to</strong><br />

1043-4666/$ - see front matter Ó 2008 Published by Elsevier Ltd.<br />

doi:10.1016/j.cy<strong>to</strong>.2008.07.009<br />

Please cite this article in press as: Koyama S et al., <strong>Innate</strong> <strong>immune</strong> <strong>response</strong> <strong>to</strong> <strong>viral</strong> <strong>infection</strong>, Cy<strong>to</strong>kine (2008), doi:10.1016/<br />

j.cy<strong>to</strong>.2008.07.009


ARTICLE IN PRESS<br />

2 S. Koyama et al. / Cy<strong>to</strong>kine xxx (2008) xxx–xxx<br />

virus <strong>infection</strong>. TLR3 is expressed more widely, but is mainly expressed<br />

on conventional DCs (cDCs) [5]. The function of TLR8 is<br />

not clearly known yet.<br />

Recently endoplasmic reticulum (ER) protein UNC93B1 turned<br />

out <strong>to</strong> be essential for trafficking of TLR7 and TLR9 from ER <strong>to</strong><br />

endosome [6], but what triggers this TLR-trafficking from ER <strong>to</strong><br />

endosome before <strong>viral</strong> recognition by TLRs remains <strong>to</strong> be elucidated.<br />

A recent report suggests that TLR-sorting <strong>to</strong> the ligand<br />

may utilize au<strong>to</strong>phagy, which is a cellular process for recycling<br />

cy<strong>to</strong>solic compartments and, in the case of pDC, eliminating exogenous<br />

pathogen. Although cy<strong>to</strong>plasmic vesicular somatitis viruses<br />

(VSV) in pDC are thought <strong>to</strong> be trapped by au<strong>to</strong>phagosome<br />

expressing ATG5 and detected by TLR7 in lysosomes for a type I<br />

IFN <strong>response</strong> [7], in non-<strong>immune</strong> cells ATG5 suppresses the type<br />

I IFN <strong>response</strong> by interaction with caspase recruitment domains<br />

(CARDs) presented by RIG-I and IFN-b promoter stimula<strong>to</strong>r-1<br />

(IPS-1) [8] (Fig. 1).<br />

2.2. Endosomal recognition of <strong>viral</strong> RNA by TLRs<br />

TLR7 recognizes several kinds of RNA viruses, including<br />

orthomyxoviruses in pDCs. TLR7 signals through a TIR domaincontaining<br />

adapter, myeloid differentiation fac<strong>to</strong>r 88 (MyD88).<br />

Upon exposure <strong>to</strong> its ligand, MyD88 forms a complex with interleukin-1-recep<strong>to</strong>r<br />

(IL-1R)-associated kinase-4 (IRAK-4), IRAK-1, tumor<br />

necrosis fac<strong>to</strong>r-recep<strong>to</strong>r associated fac<strong>to</strong>r 3 (TRAF3), TRAF6,<br />

Ikka and IRF-7 [9–11]. Following the formation of this signaling<br />

complex, IRF7 and nuclear fac<strong>to</strong>r-kappa B (NF-jB) are activated,<br />

which results in the production of type I IFNs and cy<strong>to</strong>kines (Fig. 1).<br />

TLR3 recognizes double-stranded (ds)RNA and triggers a signaling<br />

pathway via a TIR domain-containing adapter inducing IFN-b<br />

(TRIF) (also known as TICAM-1) [12,13]. TRIF associates with<br />

TRAF3 and TRAF6 via TRAF-binding motifs which exist in its N-terminal<br />

portion and also with recep<strong>to</strong>r interacting protein (RIP) 1<br />

and RIP3 via RIP homotypic interaction motif (RHIM) [14,15].<br />

TRAF6 and RIP1 activate NF-jB while TRAF3 activates TRAF family<br />

member-associated NK-jB activa<strong>to</strong>r (TANK)-binding kinase 1<br />

(TBK1) and inducible IjB kinase (IKK-i). Activation of these pathways<br />

triggers anti<strong>viral</strong> <strong>response</strong>s (Fig. 1). TLR3 also activates the<br />

phosphatidylinosi<strong>to</strong>l-3 kinase (PI3K) pathway [16]. Tyrosine phosphorylation<br />

of TLR3 induces PI3K recruitment <strong>to</strong> the recep<strong>to</strong>r and<br />

subsequent activation of Akt leads <strong>to</strong> activation of IRF-3. TLR3<br />

plays an important role in the pathogenesis of RNA virus <strong>infection</strong>s<br />

in vivo. For example, TLR3 deficient mice are resistant <strong>to</strong> <strong>infection</strong><br />

Fig. 1. RNA sensing in virus <strong>infection</strong>. TLR3 recognizes dsRNA and triggers a signaling pathway via a TRIF. TRIF associates with TRAF3, TRAF6 and RIP1. TRAF6 and RIP1<br />

activate NF-kB and AP-1 while TRAF3 activates TBK1/IKK-i and is followed by a type I IFN <strong>response</strong>. Both RIG-I and MDA5 associate with an adapter protein IPS-1. IPS-1<br />

localizes on the outer mi<strong>to</strong>chondrial membrane and the CARD of it interacts with that of RIG-I or MDA5. IPS-1 associates with TRAF3 which induces the production of type I<br />

IFNs and FADD which induces activation of NF-kB. TLR7 signals through MyD88. MyD88 forms a complex with IRAK-4, IRAK-1, TRAF3, TRAF6, Ikka and IRF-7 and this complex<br />

is recruited <strong>to</strong> the TLR by ligand stimulation. Downstream of this signaling complex, IRF7 and NF-kB are activated and this is followed by the production of type I IFNs and<br />

cy<strong>to</strong>kines. ER protein UNC93B1 plays a key role in trafficking of TLR7 from ER <strong>to</strong> endosome, but the triggers are unknown. In the case of VSV recognition in pDC, the virus is<br />

trapped by au<strong>to</strong>phagosome expressing ATG5 and detected by TLR7 in the lysosome for production of a type I IFN <strong>response</strong>. Moreover in non-<strong>immune</strong> cells ATG5 suppresses<br />

the type I IFN <strong>response</strong> via RIG-I or IPS-1 inhibition. Yellow rectangles (TIR, CARD) indicate protein–protein interaction regions for downstream signaling.<br />

Please cite this article in press as: Koyama S et al., <strong>Innate</strong> <strong>immune</strong> <strong>response</strong> <strong>to</strong> <strong>viral</strong> <strong>infection</strong>, Cy<strong>to</strong>kine (2008), doi:10.1016/<br />

j.cy<strong>to</strong>.2008.07.009


ARTICLE IN PRESS<br />

S. Koyama et al. / Cy<strong>to</strong>kine xxx (2008) xxx–xxx 3<br />

with West Nile virus [17] and influenza virus [18]. In both cases<br />

inflamma<strong>to</strong>ry <strong>response</strong>s are decreased in TLR3 deficient mice,<br />

which suggests that an excess production of cy<strong>to</strong>kines is rather<br />

harmful for the survival of mice.<br />

2.3. Intracellular recognition of <strong>viral</strong> RNA by RLRs<br />

Recently, three homologous DExD/H box RNA helicases, RIG-I,<br />

melanoma differentiation-associated gene 5 (MDA5) and LGP2,<br />

were identified as cy<strong>to</strong>plasmic sensors of virus RNA, named here<br />

as RIG-like recep<strong>to</strong>rs (RLRs). RIG-I and MDA5 play a major role in<br />

recognition of RNA viruses in cDCs, macrophages and fibroblasts.<br />

RIG-I and MDA5 share two N-terminal CARDs followed by an<br />

RNA helicase domain [19] while LGP2 lacks a CARD. RIG-I binds<br />

5 0 -triphosphorylated single-stranded (ss)RNA [20] and short<br />

dsRNA [21] and stimulates production of type I IFNs. By contrast,<br />

MDA5 preferentially recognizes longer-dsRNA, including synthetic<br />

poly-IC [22]. RIG-I recognizes a variety of RNA viruses<br />

including influenza virus, VSV and Japanese encephalitis virus<br />

(JEV) while MDA5 recognizes picorna family such as encephalomyocarditis<br />

virus (EMCV), Theiler’s virus and Mengo virus [22].<br />

Therefore, RIG-I and MDA5 deficient mice are highly susceptible<br />

<strong>to</strong> VSV and EMCV, respectively. In addition it was indicated that<br />

the anti<strong>viral</strong> protein RNase L, which can cleave and turn a singlestranded<br />

portion of not only <strong>viral</strong> but also self RNA in<strong>to</strong> preferentially<br />

double-stranded form RNA, is a ligand for RIG-I and<br />

MDA5 [23].<br />

The CARDs of RIG-I and MDA5 are responsible for initiating the<br />

signaling pathway. Both RIG-I and MDA5 associate with an adapter<br />

protein IPS-1 also known as MAVS, VISA or CARDIF, which also<br />

contains an N-terminal CARD [24–27]. IPS-1 localizes on the outer<br />

mi<strong>to</strong>chondrial membrane. The IPS-1 CARD interacts with that of<br />

RIG-I or MDA5. IPS-1 then associates with TRAF3, followed by activation<br />

of TBK1 and Ikk-i. These kinases phosphorylate IRF3 and<br />

IRF7 and induce type I IFN production [28,29]. IPS-1 also interacts<br />

with Fas-associated death domain-containing protein (FADD) and<br />

leads <strong>to</strong> activation of NF-jB through cleavage of caspase-8/-10<br />

[30] (Fig. 1).<br />

2.4. Differential role of TLR and RLR in anti<strong>viral</strong> <strong>response</strong>s<br />

Occasionally both TLR and RLR are engaged for sensing the same<br />

dsRNA or ssRNA. Normally dsRNA does not exist in host cells but in<br />

virus <strong>infection</strong> it is detected as not only a <strong>viral</strong> structure but also as<br />

a byproduct of <strong>viral</strong> replication. dsRNA activates macrophages and<br />

dendritic cells via TLR3 <strong>to</strong> secrete pro-inflamma<strong>to</strong>ry cy<strong>to</strong>kines,<br />

especially IL-12. However, type-I IFNs are produced by virus-infected<br />

cells such as fibroblasts by TLR3 independently. MDA5 recognizes<br />

synthetic dsRNA, poly-IC, and the ssRNA virus, EMCV,<br />

which generates dsRNA during replication, and induces the type-I<br />

IFN <strong>response</strong> [22,31]. Poly-IC is neither capable of inducing an innate<br />

<strong>immune</strong> <strong>response</strong> nor of working as an adjuvant in TRIF/<br />

IPS-1 double knockout mice [32].<br />

In contrast <strong>to</strong> dsRNA, ssRNA abundantly exists not only in<br />

pathogens but also in host cells. ssRNA is recognized by TLR7<br />

(or TLR8 in humans) and RIG-I in a cell-type specific manner.<br />

Although TLR7 and TLR8 recognize GU of AU rich sequences of<br />

ssRNA viruses such as influenza virus and HIV, through TLR7<br />

expressing cells such as pDC, or TLR8 expressing cells such as<br />

myeloid DC or monocytes, it is unclear whether its sequence<br />

specificity is dependent on the recep<strong>to</strong>r or the cell [33]. In contrast<br />

<strong>to</strong> TLR7, RIG-I is expressed in most cell types. As described<br />

previously, RIG-I recognizes 5 0 -triphosphorylated ssRNA [20]. In<br />

the case of influenza A virus <strong>infection</strong>, its negative-sense ssRNA<br />

genome is recognized by TLR7 expressed in pDCs and a signal is<br />

transmitted through its adap<strong>to</strong>r protein MyD88 [34]. On the other<br />

hand, it is recognized by RIG-I ubiqui<strong>to</strong>usly expressed in most cell<br />

types, such as fibroblasts or cDCs in vitro [35], and probably by<br />

the alveolar macrophage in vivo [36], via its adap<strong>to</strong>r protein<br />

IPS-1. In mouse lungs after intranasal <strong>infection</strong> of influenza virus<br />

both TLR7/MyD88 and RIG-I/IPS-1 pathways concurrently control<br />

the type-I IFN <strong>response</strong> [37].<br />

2.5. Endosomal and intracellular recognition of <strong>viral</strong> DNA<br />

TLR9 recognizes unmethylated DNA with a CpG motif (CpG-<br />

DNA) and DNA viruses, including herpes simplex virus (HSV)-1,<br />

HSV-2 and cy<strong>to</strong>megalovirus (CMV) in pDCs. TLR9 shares the adapter<br />

protein MyD88 and the downstream signaling pathway with<br />

TLR7. cDCs and macrophages also respond <strong>to</strong> CpG-DNA and produce<br />

small amounts of IFN-b through IRF-1 activation rather than<br />

IRF-3 or IRF-7 activation [38].<br />

Recently, it was reported that genomic DNA of viruses, such as<br />

adenovirus, vaccinia virus and HSV [39–41], could be also recognized<br />

in a TLR9-independent manner, using an as yet unknown<br />

recognition mechanism in the cy<strong>to</strong>plasm [42,43]. In this case,<br />

DNA which has entered the cy<strong>to</strong>plasm activates the infected cells<br />

via TBK1 and IRF3 [44]. Actually the source of DNA is not restricted<br />

<strong>to</strong> viruses; but it can also come from bacteria and damaged host<br />

cells. The activity of this DNA is more potent in ds right-hand B-<br />

form DNA than in left-handed Z-form DNA, while ssDNA displays<br />

no activity [40,44,45] (Fig. 2). In addition DAI (also known as<br />

ZBP1 or DLM1) which contains two Z-DNA binding domains, was<br />

shown <strong>to</strong> be a potential cy<strong>to</strong>plasmic DNA sensor [46]. However,<br />

DAI KO mice induced a normal type I IFN <strong>response</strong> in vitro and<br />

in vivo after B-DNA stimulation and also indicated DNA-vaccine-induced<br />

adaptive <strong>immune</strong> <strong>response</strong>s, suggesting its role is redundant<br />

[47]. Potential cy<strong>to</strong>plasmic DNA sensors still remain <strong>to</strong> be<br />

elucidated.<br />

2.6. The recognition of <strong>viral</strong> components at the cell surface<br />

In addition <strong>to</strong> the endosomal TLRs, TLR2 and TLR4 have also<br />

been suggested <strong>to</strong> be involved in recognition of viruses. TLR2 has<br />

been shown <strong>to</strong> detect components of measles virus, HSV and hepatitis<br />

C virus (HCV) [48–50], while TLR4 produces a <strong>response</strong> <strong>to</strong> retrovirus<br />

and respira<strong>to</strong>ry syncytial virus (RSV) [51,52]. While <strong>viral</strong><br />

proteins recognized by these surface TLRs trigger pro-inflamma<strong>to</strong>ry<br />

<strong>response</strong>s, their contribution <strong>to</strong> either protective or pathological<br />

<strong>immune</strong> <strong>response</strong>s largely depends on the type of virus, route<br />

of <strong>infection</strong>, and other host fac<strong>to</strong>rs [53].<br />

2.7. NLRs mediates innate <strong>immune</strong> activation by intracellular <strong>viral</strong><br />

nucleic acids<br />

NLR proteins are comprised three motifs, C-terminal LRRs, central<br />

nucleotide-binding domain and N-terminal signaling domaincontaining<br />

CARDs, and Pyrin domain or baculovirus IAP repeats<br />

[54]. Cryopyrin/NALP3 was shown <strong>to</strong> recognize both ssRNA and<br />

dsRNA of <strong>viral</strong> origin or their synthetic versions and <strong>to</strong> induce caspase-1<br />

activation via apop<strong>to</strong>sis-associated speck-like protein containing<br />

a caspase-activating and recruitment domain (ASC)<br />

[55,56]. In addition, some NLRs participate in nucleic acid-mediated<br />

innate <strong>immune</strong> activation through caspase-1 activation<br />

[56,57], and NF-jB activation <strong>to</strong>wards IFN-I production via a synergistic<br />

pathway activated by NOD2 [58].<br />

3. Discrimination between self and <strong>viral</strong> nucleic acids<br />

The innate <strong>immune</strong> systems for virus sensing described above<br />

miraculously detect the invasion of pathogens such as viruses,<br />

Please cite this article in press as: Koyama S et al., <strong>Innate</strong> <strong>immune</strong> <strong>response</strong> <strong>to</strong> <strong>viral</strong> <strong>infection</strong>, Cy<strong>to</strong>kine (2008), doi:10.1016/<br />

j.cy<strong>to</strong>.2008.07.009


ARTICLE IN PRESS<br />

4 S. Koyama et al. / Cy<strong>to</strong>kine xxx (2008) xxx–xxx<br />

Fig. 2. DNA sensing in virus <strong>infection</strong>. TLR9 recognizes CpG-DNA and DNA viruses, including HSV-1, HSV-2 and CMV. TLR9 shares MyD88 and a downstream signaling<br />

pathway with TLR7 and ER protein UNC93B1 also plays a key role in trafficking of TLR9 from ER <strong>to</strong> endosome. In TLR9-independent DNA sensing, DNAs enter in<strong>to</strong> the<br />

cy<strong>to</strong>plasm which activates the infected cells via TBK1 and IRF3 but the recep<strong>to</strong>r and adap<strong>to</strong>r involved are still unknown. Their activities are more potent in ds right-hand B-<br />

form DNA than in left-handed Z-form DNA.<br />

but are silent in normal conditions. As dsRNA generated by <strong>viral</strong><br />

replication and virus DNA rich in CpG motifs are not normally<br />

found in our body, it is easy <strong>to</strong> consider that our innate <strong>immune</strong><br />

system recognizes them as foreign molecules. In contrast <strong>to</strong> these<br />

nucleic acids, ssRNA abundantly exists not only in pathogens but<br />

also in host cells. It is, therefore, essential for the host <strong>to</strong> detect<br />

and discriminate <strong>viral</strong> ssRNA from self ssRNA. However, the mechanism<br />

of this robust discrimination is not fully unders<strong>to</strong>od. For<br />

example, although the 5 0 triphosphate on many ssRNAs of viruses<br />

is absent from mRNA and transfer RNA but is found in ribosomal<br />

RNA, which abundantly exists in host cells, only the 5 0 triphosphate<br />

on viruses can induce RIG-I activation [59,60]. Recently RNase L<br />

activation in infected host cells was shown <strong>to</strong> generate small self<br />

RNAs which can induce an innate <strong>immune</strong> <strong>response</strong> via RLRs<br />

[61]. In addition, it is known that TLR7/8 and TLR9 also recognize<br />

host RNA and DNA. Therefore, it is necessary for absolute discrimination<br />

by the host innate <strong>immune</strong> system <strong>to</strong> recognize some additional<br />

fac<strong>to</strong>rs such as the methylation state, certain sequences and<br />

intracellular localization of RNA, or restricted endosomal expression<br />

of TLRs for <strong>viral</strong> recognition where host nucleic acids have limited<br />

access [62–64].<br />

4. Virulent fac<strong>to</strong>r for inhibition of host <strong>immune</strong> <strong>response</strong><br />

Viruses have developed several kinds of <strong>immune</strong> evasion strategies<br />

<strong>to</strong> proliferate within host cells. Their main target is the type I<br />

IFN <strong>response</strong>. Viruses can inhibit type I IFNs by many strategies i.e.<br />

inhibition of IFN synthesis, interference of IFN recep<strong>to</strong>r signaling<br />

and so on [65]. For example, vaccinia virus E3L and influenza virus<br />

NS1 which possess a dsRNA-binding site are thought <strong>to</strong> inhibit<br />

type I IFN production through dsRNA sequestration [66,67]. As<br />

E3L also possesses a DNA-binding site, it might sequester <strong>viral</strong><br />

DNA from host DNA sensing [68]. NS1 protein was also indicated<br />

<strong>to</strong> inhibit the function of IPS1 and RIG-I [69]. Viruses without such<br />

abilities <strong>to</strong> suppress the host type I IFN <strong>response</strong>s are generally low<br />

pathogenic and available for vaccine strains.<br />

5. Adaptive immunity against viruses through innate <strong>immune</strong><br />

signaling pathway<br />

Recent advances in the understanding of innate immunity show<br />

that the activation of the innate <strong>immune</strong> system is essential for<br />

subsequent adaptive <strong>immune</strong> <strong>response</strong>s including specific antibody<br />

production and CTL activation which play a key role in protection<br />

against virus <strong>infection</strong>. A recent report indicated that the<br />

adaptive <strong>immune</strong> <strong>response</strong> elicited by inactivated whole influenza<br />

virus vaccine containing <strong>viral</strong> ssRNA was strictly governed by the<br />

TLR7/MyD88 pathway, but not by the RIG-I/IPS-1 pathway,<br />

although both pathways concurrently controlled the innate <strong>immune</strong><br />

<strong>response</strong> [37]. However, the innate <strong>immune</strong> <strong>response</strong> elicited<br />

by a DNA vaccine containing CpG-DNA was dependent on<br />

TBK1, but not on the TLR9/MyD88 pathway [47]. These results sug-<br />

Please cite this article in press as: Koyama S et al., <strong>Innate</strong> <strong>immune</strong> <strong>response</strong> <strong>to</strong> <strong>viral</strong> <strong>infection</strong>, Cy<strong>to</strong>kine (2008), doi:10.1016/<br />

j.cy<strong>to</strong>.2008.07.009


ARTICLE IN PRESS<br />

S. Koyama et al. / Cy<strong>to</strong>kine xxx (2008) xxx–xxx 5<br />

gested that the innate <strong>immune</strong> pathway engaged for protective<br />

immunity against virus <strong>infection</strong> was different according <strong>to</strong> the<br />

source of antigen.<br />

6. Conclusion<br />

This review has illustrated the recent progress in understanding<br />

how a host discriminates between viruses and self components by<br />

innate <strong>immune</strong> recep<strong>to</strong>rs and elicits an inflamma<strong>to</strong>ry <strong>response</strong><br />

involving type I IFNs and other cy<strong>to</strong>kines. Much remains <strong>to</strong> be clarified<br />

about the complex interplay between host and virus, but elucidating<br />

such mechanisms in detail is essential for not only the<br />

development of a clinical approach such as nucleic acid-based<br />

immunotherapy and TLR based vaccine adjuvant but also the<br />

understanding of the pathogenesis of diverse <strong>viral</strong> diseases.<br />

Acknowledgments<br />

We thank members of Akira Labora<strong>to</strong>ry, Prof. Toshihiro Horii<br />

and his labora<strong>to</strong>ry members for discussions and contributions <strong>to</strong><br />

the work discussed here. This work was supported by grants from<br />

the Ministry of Education, Culture, Sports, Science and Technology<br />

in Japan.<br />

References<br />

[1] Stetson DB, Medzhi<strong>to</strong>v R. Type I interferons in host defense. Immunity<br />

2006;25:373–81.<br />

[2] Kotenko SV, Gallagher G, Baurin VV, Lewis-Antes A, Shen M, Shah NK, et al.<br />

IFN-lambdas mediate anti<strong>viral</strong> protection through a distinct class II cy<strong>to</strong>kine<br />

recep<strong>to</strong>r complex. Nat Immunol 2003;4:69–77.<br />

[3] Samuel CE. Anti<strong>viral</strong> actions of interferons. Clin Microbiol Rev<br />

2001;14:778–809.<br />

[4] Le Bon A, Tough DF. Links between innate and adaptive immunity via type I<br />

interferon. Curr Opin Immunol 2002;14:432–6.<br />

[5] Iwasaki A, Medzhi<strong>to</strong>v R. Toll-like recep<strong>to</strong>r control of the adaptive <strong>immune</strong><br />

<strong>response</strong>s. Nat Immunol 2004;5:987–95.<br />

[6] Kim YM, Brinkmann MM, Paquet ME, Ploegh HL. UNC93B1 delivers<br />

nucleotide-sensing <strong>to</strong>ll-like recep<strong>to</strong>rs <strong>to</strong> endolysosomes. Nature<br />

2008;452:234–8.<br />

[7] Lee HK, Lund JM, Ramanathan B, Mizushima N, Iwasaki A. Au<strong>to</strong>phagydependent<br />

<strong>viral</strong> recognition by plasmacy<strong>to</strong>id dendritic cells. Science<br />

2007;315:1398–401.<br />

[8] Jounai N, Takeshita F, Kobiyama K, Sawano A, Miyawaki A, Xin KQ, et al. The<br />

Atg5–Atg12 conjugate associates with innate anti<strong>viral</strong> <strong>immune</strong> <strong>response</strong>s. Proc<br />

Natl Acad Sci USA 2007;104:14050–5.<br />

[9] Häcker H, Redecke V, Blagoev B, Kratchmarova I, Hsu LC, Wang GG, et al.<br />

Specificity in Toll-like recep<strong>to</strong>r signalling through distinct effec<strong>to</strong>r functions of<br />

TRAF3 and TRAF6. Nature 2006;439:204–7.<br />

[10] Kawai T, Sa<strong>to</strong> S, Ishii KJ, Coban C, Hemmi H, Yamamo<strong>to</strong> M, et al. Interferonalpha<br />

induction through Toll-like recep<strong>to</strong>rs involves a direct interaction of<br />

IRF7 with MyD88 and TRAF6. Nat Immunol 2004;5:1061–8.<br />

[11] Uematsu S, Sa<strong>to</strong> S, Yamamo<strong>to</strong> M, Hirotani T, Ka<strong>to</strong> H, Takeshita F, et al.<br />

Interleukin-1 recep<strong>to</strong>r-associated kinase-1 plays an essential role for Toll-like<br />

recep<strong>to</strong>r (TLR)-7 and TLR9-mediated interferon-alpha induction. J Exp Med<br />

2005;201:915–23.<br />

[12] Yamamo<strong>to</strong> M, Sa<strong>to</strong> S, Mori K, Hoshino K, Takeuchi O, Takeda K, et al. Cutting<br />

edge: a novel Toll/IL-1 recep<strong>to</strong>r domain-containing adapter that preferentially<br />

activates the IFN-beta promoter in the Toll-like recep<strong>to</strong>r signaling. J Immunol<br />

2002;169:6668–72.<br />

[13] Oshiumi H, Matsumo<strong>to</strong> M, Funami K, Akazawa T, Seya T. TICAM-1, an adap<strong>to</strong>r<br />

molecule that participates in Toll-like recep<strong>to</strong>r 3-mediated interferon-beta<br />

induction. Nat Immunol 2003;4:161–7.<br />

[14] Sa<strong>to</strong> S, Sugiyama M, Yamamo<strong>to</strong> M, Watanabe Y, Kawai T, Takeda K, et al.<br />

Toll/IL-1 recep<strong>to</strong>r domain-containing adap<strong>to</strong>r inducing IFN-beta (TRIF)<br />

associates with TNF recep<strong>to</strong>r-associated fac<strong>to</strong>r 6 and TANK-binding kinase<br />

1, and activates two distinct transcription fac<strong>to</strong>rs, NF-kappa B and IFNregula<strong>to</strong>ry<br />

fac<strong>to</strong>r-3, in the Toll-like recep<strong>to</strong>r signaling. J Immunol<br />

2003;171:4304–10.<br />

[15] Meylan E, Burns K, Hofmann K, Blancheteau V, Martinon F, Kelliher M, et al.<br />

RIP1 is an essential media<strong>to</strong>r of Toll-like recep<strong>to</strong>r 3-induced NF-kappa B<br />

activation. Nat Immunol 2004;5:503–7.<br />

[16] Sarkar SN, Peters KL, Elco CP, Sakamo<strong>to</strong> S, Pal S, Sen GC. Novel roles of TLR3<br />

tyrosine phosphorylation and PI3 kinase in double-stranded RNA signaling.<br />

Nat Struct Mol Biol 2004;11:1060–7.<br />

[17] Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E, Flavell RA. Toll-like<br />

recep<strong>to</strong>r 3 mediates West Nile virus entry in<strong>to</strong> the brain causing lethal<br />

encephalitis. Nat Med 2004;10:1366–73.<br />

[18] Le Goffic R, Balloy V, Lagranderie M, Alexopoulou L, Escriou N, Flavell R, et al.<br />

Detrimental contribution of the Toll-like recep<strong>to</strong>r (TLR)3 <strong>to</strong> influenza A virusinduced<br />

acute pneumonia. PLoS Pathog 2006;2:e53.<br />

[19] Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T, Miyagishi<br />

M, et al. The RNA helicase RIG-I has an essential function in doublestranded<br />

RNA-induced innate anti<strong>viral</strong> <strong>response</strong>s. Nat Immunol<br />

2004;5:730–7.<br />

[20] Pichlmair A, Schulz O, Tan CP, Naslund TI, Liljestrom P, Weber F, et al. RIG-Imediated<br />

anti<strong>viral</strong> <strong>response</strong>s <strong>to</strong> single-stranded RNA bearing 5 0 -phosphates.<br />

Science 2006;314:997–1001.<br />

[21] Takahasi K, Yoneyama M, Nishihori T, Hirai R, Kumeta H, Narita R, et al.<br />

Nonself RNA-sensing mechanism of RIG-I helicase and activation of anti<strong>viral</strong><br />

<strong>immune</strong> <strong>response</strong>s. Mol Cell 2008;29:428–40.<br />

[22] Ka<strong>to</strong> H, Takeuchi O, Sa<strong>to</strong> S, Yoneyama M, Yamamo<strong>to</strong> M, Matsui K, et al.<br />

Differential roles of MDA5 and RIG-I helicases in the recognition of RNA<br />

viruses. Nature 2006;441:101–5.<br />

[23] Malathi K, Dong B, Gale Jr M, Silverman RH. Small self-RNA generated by<br />

RNase L amplifies anti<strong>viral</strong> innate immunity. Nature 2007;448:816–9.<br />

[24] Kawai T, Takahashi K, Sa<strong>to</strong> S, Coban C, Kumar H, Ka<strong>to</strong> H, et al. IPS-1, an adap<strong>to</strong>r<br />

triggering RIG-I- and Mda5-mediated type I interferon induction. Nat<br />

Immunol 2005;6:981–8.<br />

[25] Seth RB, Sun L, Ea CK, Chen ZJ. Identification and characterization of MAVS, a<br />

mi<strong>to</strong>chondrial anti<strong>viral</strong> signaling protein that activates NF-kappaB and IRF 3.<br />

Cell 2005;122:669–82.<br />

[26] Meylan E, Curran J, Hofmann K, Moradpour D, Binder M, Bartenschlager R,<br />

et al. Cardif is an adap<strong>to</strong>r protein in the RIG-I anti<strong>viral</strong> pathway and is targeted<br />

by hepatitis C virus. Nature 2005;437:1167–72.<br />

[27] Xu LG, Wang YY, Han KJ, Li LY, Zhai Z, Shu HB. VISA is an adapter protein<br />

required for virus-triggered IFN-beta signaling. Mol Cell 2005;19:727–40.<br />

[28] Hemmi H, Takeuchi O, Sa<strong>to</strong> S, Yamamo<strong>to</strong> M, Kaisho T, Sanjo H, et al. The<br />

roles of two IkappaB kinase-related kinases in lipopolysaccharide and<br />

double stranded RNA signaling and <strong>viral</strong> <strong>infection</strong>. J Exp Med<br />

2004;199:1641–50.<br />

[29] Honda K, Yanai H, Negishi H, Asagiri M, Sa<strong>to</strong> M, Mizutani T, et al. IRF-7 is the<br />

master regula<strong>to</strong>r of type-I interferon-dependent <strong>immune</strong> <strong>response</strong>s. Nature<br />

2005;434:772–7.<br />

[30] Takahashi K, Kawai T, Kumar H, Sa<strong>to</strong> S, Yonehara S, Akira S. Roles of caspase-8<br />

and caspase-10 in innate <strong>immune</strong> <strong>response</strong>s <strong>to</strong> double-stranded RNA. J<br />

Immunol 2006;176:4520–4.<br />

[31] Gitlin L, Barchet W, Gilfillan S, Cella M, Beutler B, Flavell RA, et al. Essential role<br />

of mda-5 in type I IFN <strong>response</strong>s <strong>to</strong> polyriboinosinic:polyribocytidylic acid and<br />

encephalomyocarditis picornavirus. Proc Natl Acad Sci USA<br />

2006;103:8459–64.<br />

[32] Kumar H, Koyama S, Ishii KJ, Kawai T, Akira S. Cutting edge: cooperation of IPS-<br />

1- and TRIF-dependent pathways in poly IC-enhanced antibody production<br />

and cy<strong>to</strong><strong>to</strong>xic T cell <strong>response</strong>s. J Immunol 2008;180:683–7.<br />

[33] Diebold SS. Recognition of <strong>viral</strong> single-stranded RNA by Toll-like recep<strong>to</strong>rs.<br />

Adv Drug Deliv Rev 2008;60:813–23.<br />

[34] Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. <strong>Innate</strong> anti<strong>viral</strong><br />

<strong>response</strong>s by means of TLR7-mediated recognition of single-stranded RNA.<br />

Science 2004;303:1529–31.<br />

[35] Ka<strong>to</strong> H, Sa<strong>to</strong> S, Yoneyama M, Yamamo<strong>to</strong> M, Uematsu S, Matsui K, et al. Cell<br />

type-specific involvement of RIG-I in anti<strong>viral</strong> <strong>response</strong>. Immunity<br />

2005;23:19–28.<br />

[36] Kumagai Y, Takeuchi O, Ka<strong>to</strong> H, Kumar H, Matsui K, Morii E, et al. Alveolar<br />

macrophages are the primary interferon-alpha producer in pulmonary<br />

<strong>infection</strong> with RNA viruses. Immunity 2007;27:240–52.<br />

[37] Koyama S, Ishii KJ, Kumar H, Tanimo<strong>to</strong> T, Coban C, Uematsu S, et al. Differential<br />

role of TLR- and RLR-signaling in the <strong>immune</strong> <strong>response</strong>s <strong>to</strong> influenza A virus<br />

<strong>infection</strong> and vaccination. J Immunol 2007;179:4711–20.<br />

[38] Schmitz F, Heit A, Guggemoos S, Krug A, Mages J, Schiemann M, et al.<br />

Interferon-regula<strong>to</strong>ry-fac<strong>to</strong>r 1 controls Toll-like recep<strong>to</strong>r 9-mediated IFN-beta<br />

production in myeloid dendritic cells. Eur J Immunol. 2007;37:315–27.<br />

[39] Nociari M, Ocheretina O, Schoggins JW, Falck-Pedersen E. Sensing <strong>infection</strong> by<br />

adenovirus: Toll-like recep<strong>to</strong>r-independent <strong>viral</strong> DNA recognition signals<br />

activation of the interferon regula<strong>to</strong>ry fac<strong>to</strong>r 3 master regula<strong>to</strong>r. J Virol<br />

2007;81:4145–57.<br />

[40] Ishii KJ, Coban C, Ka<strong>to</strong> H, Takahashi K, Torii Y, Takeshita F, et al. A Toll-like<br />

recep<strong>to</strong>r-independent anti<strong>viral</strong> <strong>response</strong> induced by double-stranded B-form<br />

DNA. Nat Immunol 2006;7:40–8.<br />

[41] Hochrein H, Schlatter B, O’Keeffe M, Wagner C, Schmitz F, Schiemann M, et al.<br />

Herpes simplex virus type-1 induces IFN-alpha production via Toll-like<br />

recep<strong>to</strong>r 9-dependent and -independent pathways. Proc Natl Acad Sci USA<br />

2004;101:11416–21.<br />

[42] Wagner H, Bauer S. All is not Toll: new pathways in DNA recognition. J Exp<br />

Med 2006;203:265–8.<br />

[43] Ishii KJ, Akira S. <strong>Innate</strong> <strong>immune</strong> recognition of, and regulation by, DNA. Trends<br />

Immunol 2006;27:525–32.<br />

[44] Stetson DB, Medzhi<strong>to</strong>v R. Recognition of cy<strong>to</strong>solic DNA activates an IRF3-<br />

dependent innate <strong>immune</strong> <strong>response</strong>. Immunity 2006;24:93–103.<br />

[45] Okabe Y, Kawane K, Akira S, Taniguchi T, Nagata S. Toll-like recep<strong>to</strong>rindependent<br />

gene induction program activated by mammalian DNA escaped<br />

from apop<strong>to</strong>tic DNA degradation. J Exp Med 2005;202:1333–9.<br />

[46] Takaoka A, Wang Z, Choi MK, Yanai H, Negishi H, Ban T, et al. DAI (DLM-1/<br />

ZBP1) is a cy<strong>to</strong>solic DNA sensor and an activa<strong>to</strong>r of innate <strong>immune</strong> <strong>response</strong>.<br />

Nature 2007;448:501–5.<br />

Please cite this article in press as: Koyama S et al., <strong>Innate</strong> <strong>immune</strong> <strong>response</strong> <strong>to</strong> <strong>viral</strong> <strong>infection</strong>, Cy<strong>to</strong>kine (2008), doi:10.1016/<br />

j.cy<strong>to</strong>.2008.07.009


ARTICLE IN PRESS<br />

6 S. Koyama et al. / Cy<strong>to</strong>kine xxx (2008) xxx–xxx<br />

[47] Ishii KJ, Kawagoe T, Koyama S, Matsui K, Kumar H, Kawai T, et al. TANKbinding<br />

kinase-1 delineates innate and adaptive <strong>immune</strong> <strong>response</strong>s <strong>to</strong> DNA<br />

vaccines. Nature 2008;451:725–9.<br />

[48] Bieback K, Lien E, Klagge IM, Avota E, Schneider-Schaulies J, Duprex WP, et al.<br />

Hemagglutinin protein of wild-type measles virus activates <strong>to</strong>ll-like recep<strong>to</strong>r 2<br />

signaling. J Virol 2002;76:8729–36.<br />

[49] Kurt-Jones EA, Chan M, Zhou S, Wang J, Reed G, Bronson R, et al. Herpes<br />

simplex virus 1 interaction with Toll-like recep<strong>to</strong>r 2 contributes <strong>to</strong> lethal<br />

encephalitis. Proc Natl Acad Sci USA 2004;101:1315–20.<br />

[50] Duesberg U, Von dem Bussche A, Kirschning C, Miyake K, Sauerbruch T,<br />

Spengler U. Cell activation by synthetic lipopeptides of the hepatitis C virus<br />

(HCV)–core protein is mediated by <strong>to</strong>ll like recep<strong>to</strong>rs (TLRs) 2 and 4. Immunol<br />

Lett 2002;84:89–95.<br />

[51] Kurt-Jones EA, Popova L, Kwinn L, Haynes LM, Jones LP, Tripp RA, et al. Pattern<br />

recognition recep<strong>to</strong>rs TLR4 and CD14 mediate <strong>response</strong> <strong>to</strong> respira<strong>to</strong>ry<br />

syncytial virus. Nat Immunol 2000;1:398–401.<br />

[52] Rassa JC, Meyers JL, Zhang Y, Kudaravalli R, Ross SR. Murine retroviruses<br />

activate B cells via interaction with <strong>to</strong>ll-like recep<strong>to</strong>r 4. Proc Natl Acad Sci USA<br />

2002;99:2281–6.<br />

[53] Finberg RW, Wang JP, Kurt-Jones EA. Toll like recep<strong>to</strong>rs and viruses. Rev Med<br />

Virol 2007;17:35–43.<br />

[54] Meylan E, Tschopp J, Karin M. Intracellular pattern recognition recep<strong>to</strong>rs in the<br />

host <strong>response</strong>. Nature 2006;442:39–44.<br />

[55] Kanneganti TD, Body-Malapel M, Amer A, Park JH, Whitfield J, Franchi L, et al.<br />

Critical role for Cryopyrin/Nalp3 in activation of caspase-1 in <strong>response</strong> <strong>to</strong> <strong>viral</strong><br />

<strong>infection</strong> and double-stranded RNA. J Biol Chem 2006;281:36560–8.<br />

[56] Kanneganti TD, Ozoren N, Body-Malapel M, Amer A, Park JH, Franchi L, et al.<br />

Bacterial RNA and small anti<strong>viral</strong> compounds activate caspase-1 through<br />

cryopyrin/Nalp3. Nature 2006;440:233–6.<br />

[57] Muruve DA, Petrilli V, Zaiss AK, White LR, Clark SA, Ross PJ, et al. The<br />

inflammasome recognizes cy<strong>to</strong>solic microbial and host DNA and triggers an<br />

innate <strong>immune</strong> <strong>response</strong>. Nature 2008;452:103–7.<br />

[58] Leber JH, Crimmins GT, Raghavan S, Meyer-Morse NP, Cox JS, Portnoy DA.<br />

Distinct TLR- and NLR-mediated transcriptional <strong>response</strong>s <strong>to</strong> an intracellular<br />

pathogen. PLoS Pathog 2008;4:e6.<br />

[59] Hornung V, Ellegast J, Kim S, Brzozka K, Jung A, Ka<strong>to</strong> H, et al. 5 0 -Triphosphate<br />

RNA is the ligand for RIG-I. Science 2006;314:994–7.<br />

[60] Ramakrishnan V. Ribosome structure and the mechanism of translation. Cell<br />

2002;108:557–72.<br />

[61] Malathi K, Dong B, Gale M, Silverman R. Small self-RNA generated by RNase L<br />

amplifies anti<strong>viral</strong> innate immunity. Nature 2007;448:816–9.<br />

[62] Kariko K, Buckstein M, Ni H, Weissman D. Suppression of RNA recognition by<br />

Toll-like recep<strong>to</strong>rs: the impact of nucleoside modification and the evolutionary<br />

origin of RNA. Immunity 2005;23:165–75.<br />

[63] Ishii KJ, Akira S. TLR ignores methylated RNA? Immunity<br />

2005;23:111–3.<br />

[64] Marshak-Rothstein A, Rifkin I. Immunologically active au<strong>to</strong>antigens: the role<br />

of <strong>to</strong>ll-like recep<strong>to</strong>rs in the development of chronic inflamma<strong>to</strong>ry disease.<br />

Annu Rev Immunol 2007;25:419–41.<br />

[65] Weber F, Kochs G, Haller O. Inverse interference: how viruses fight the<br />

interferon system. Viral Immunol 2004;17:498–515.<br />

[66] Xiang Y, Condit RC, Vijaysri S, Jacobs B, Williams BR, Silverman RH. Blockade of<br />

interferon induction and action by the E3L double-stranded RNA binding<br />

proteins of vaccinia virus. J Virol 2002;76:5251–9.<br />

[67] Garcia-Sastre A. Inhibition of interferon-mediated anti<strong>viral</strong> <strong>response</strong>s by<br />

influenza A viruses and other negative-strand RNA viruses. Virology<br />

2001;279:375–84.<br />

[68] Kim YG, Muralinath M, Brandt T, Pearcy M, Hauns K, Lowenhaupt K, et al. A<br />

role for Z-DNA binding in vaccinia virus pathogenesis. Proc Natl Acad Sci USA<br />

2003;100:6974–9.<br />

[69] Mibayashi M, Martinez-Sobrido L, Loo YM, Cardenas WB, Gale Jr M, Garcia-<br />

Sastre A. Inhibition of retinoic acid-inducible gene I-mediated induction of<br />

beta interferon by the NS1 protein of influenza A virus. J Virol<br />

2007;81:514–24.<br />

Please cite this article in press as: Koyama S et al., <strong>Innate</strong> <strong>immune</strong> <strong>response</strong> <strong>to</strong> <strong>viral</strong> <strong>infection</strong>, Cy<strong>to</strong>kine (2008), doi:10.1016/<br />

j.cy<strong>to</strong>.2008.07.009

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!